Review article
CaV1.2 signaling complexes in the heart

https://doi.org/10.1016/j.yjmcc.2012.12.006Get rights and content

Abstract

L-type Ca2 + channels (LTCCs) are essential for generation of the electrical and mechanical properties of cardiac muscle. Furthermore, regulation of LTCC activity plays a central role in mediating the effects of sympathetic stimulation on the heart. The primary mechanism responsible for this regulation involves β-adrenergic receptor (βAR) stimulation of cAMP production and subsequent activation of protein kinase A (PKA). Although it is well established that PKA-dependent phosphorylation regulates LTCC function, there is still much we do not understand. However, it has recently become clear that the interaction of the various signaling proteins involved is not left to completely stochastic events due to random diffusion. The primary LTCC expressed in cardiac muscle, CaV1.2, forms a supramolecular signaling complex that includes the β2AR, G proteins, adenylyl cyclases, phosphodiesterases, PKA, and protein phosphatases. In some cases, the protein interactions with CaV1.2 appear to be direct, in other cases they involve scaffolding proteins such as A kinase anchoring proteins and caveolin-3. Functional evidence also suggests that the targeting of these signaling proteins to specific membrane domains plays a critical role in maintaining the fidelity of receptor mediated LTCC regulation. This information helps explain the phenomenon of compartmentation, whereby different receptors, all linked to the production of a common diffusible second messenger, can vary in their ability to regulate LTCC activity. The purpose of this review is to examine our current understanding of the signaling complexes involved in cardiac LTCC regulation. This article is part of a Special Issue entitled "Calcium Signaling in Heart".

Highlights

► Ca2 + influx by L-type Ca2 + channels trigger Ca2 + release and cardiac contraction. ► We discuss how VGCC are regulated by GPCR via cAMP/PKA. ► VGCC form signaling complexes with β2AR, Gs, adenylyl cyclase, and PKA. ► VGCC also associate with the protein phosphatases PP2A and PP2B. ► These complexes regulate channel activity in a highly localized manner.

Introduction

The proximity of the constituent components of a signaling pathway often plays a critical role in ensuring the speed, efficiency, and specificity of the functional responses they produce. This is especially true for the signaling mechanisms involved in regulating many different ion channels. Ion channels forming signaling complexes with kinase(s) and phosphatase(s) is a common theme [1]. While the localization of such signaling molecules is often achieved through direct protein-protein interactions, spatial organization can also be achieved indirectly via scaffolding proteins as well as the targeting of the relevant control elements to specific subcellular locations or lipid domains in the plasma membrane [1], [2], [3]. The spatial restriction of signaling also extends to aspects of those pathways that involve diffusible second messengers, such as cAMP. Accordingly, signaling complexes often include receptors and enzymes such as adenylyl cyclase that are responsible for second messenger production, as well as proteins such as phosphodiesterases (PDEs), which are involved in second messenger catabolism. The primary focus of this review will be on the signaling complexes important in maintaining the fidelity of L-type Ca2 + channel responses involving protein kinase A (PKA) in the heart.

Section snippets

Ca2 + channels in the heart

Ca2 + is a potent second messenger that controls a variety of cellular functions [4], [5]. This is particularly true in the heart, where the influx of Ca2 + through voltage-dependent Ca2 + channels plays an essential role in regulating action potential duration, triggering myocyte contraction, and controlling gene transcription [6]. Thus, they are an important target for regulating cellular function by a number of different signal transduction pathways, including those involving PKA.

Molecular structure of L-type Ca2 + channels

There are four types of LTCC, CaV1.1–1.4, each of which exists as a multimeric protein complex consisting of one of four different corresponding α1 subunits (α11.1–1.4) together with auxiliary β, α2δ, and γ subunits [7] (Fig. 1). The α1 subunit forms the ion-conducting pore and defines the specific type of Ca2 + channel. It consists of four homologous domains (I-IV) each containing six transmembrane segments (S1–S6) and a pore forming P-loop between segments 5 and 6. CaV1.2 is the predominant

Regulation of CaV1.2

The regulation of CaV1.2 has been extensively studied because of its central role in contributing to the electrical and mechanical properties of the heart. Influx of Ca2 + through LTCCs is responsible for maintaining membrane depolarization during the plateau of the cardiac action potential. Subsequent inactivation then allows repolarization thus affecting action potential duration. In this way LTCCs play a critical role in determining refractory period duration, thereby ensuring that electrical

A kinase anchoring proteins

PKA is an inactive tetramer consisting of two regulatory (inhibitory) (R) and two catalytic (C) subunits. Distinct genes encode four R (RIα, β and RIIα, β) and three C subunits (Cα, β, and γ). Suppression of C subunit catalytic activity is released following the binding of two molecules of cAMP to each R subunit [124]. It is now widely accepted that AKAPs play an essential role in orchestrating many different cAMP-dependent signaling events by tethering PKA and other regulatory enzymes together

Lipid rafts, caveolae, and cavoeolin-3

An alternative mechanism for the assembly of signaling complexes associated with CaV1.2 is through the colocalization of proteins in specific membrane domains [2]. The tight packing of sphingolipids and cholesterol in the plasma membrane creates microdomains called lipid rafts. These cholesterol rich domains can assemble signaling proteins with ion channels [140], [141], [142]. Proteins associated with lipid rafts are resistant to detergent (Triton X-100) solubilization and can be found in the

Summary

PKA-dependent phosphorylation plays an essential role in LTCC regulation. However, the exact mechanism for this regulatory effect is still an area of active investigation. Current evidence supports the idea that it may involve phosphorylation of one or more residues on α11.2, which then disrupts the autoinhibitory effect of the distal C terminal region of this subunit.

Another important aspect of LTCC regulation is the interaction of CaV1.2 with PKA. There is evidence that AKAP5 and AKAP7 can

Conflict of interest

The authors declare that there is no conflict of interest.

References (157)

  • I. Splawski et al.

    Ca(V)1.2 calcium channel dysfunction causes a multisystem disorder including arrhythmia and autism

    Cell

    (2004)
  • S.R. Agarwal et al.

    Effects of cholesterol depletion on compartmentalized cAMP responses in adult cardiac myocytes

    J Mol Cell Cardiol

    (2011)
  • R.P. Xiao et al.

    Subtype-specific beta-adrenoceptor signaling pathways in the heart and their potential clinical implications

    Trends Pharmacol Sci

    (2004)
  • R.P. Xiao et al.

    Beta 2-adrenergic receptor-stimulated increase in cAMP in rat heart cells is not coupled to changes in Ca2 + dynamics, contractility, or phospholamban phosphorylation

    J Biol Chem

    (1994)
  • S. Calaghan et al.

    Compartmentalisation of cAMP-dependent signalling by caveolae in the adult cardiac myocyte

    J Mol Cell Cardiol

    (2008)
  • Y. Chen-Izu et al.

    G(i)-dependent localization of beta(2)-adrenergic receptor signaling to L-type Ca(2 +) channels

    Biophys J

    (2000)
  • D.A. Macdougall et al.

    Caveolae compartmentalise beta2-adrenoceptor signals by curtailing cAMP production and maintaining phosphatase activity in the sarcoplasmic reticulum of the adult ventricular myocyte

    J Mol Cell Cardiol

    (2012)
  • J.S. Hayes et al.

    Selective activation of particulate cAMP-dependent protein kinase by isoproterenol and prostaglandin E1

    J Biol Chem

    (1980)
  • I.L. Buxton et al.

    Compartments of cyclic AMP and protein kinase in mammalian cardiomyocytes

    J Biol Chem

    (1983)
  • W. Schlegel et al.

    Activation of adenylate cyclase in hepatic membranes involves interactions of the catalytic unit with multimeric complexes of regulatory proteins

    J Biol Chem

    (1979)
  • M. Kuschel et al.

    G(i) protein-mediated functional compartmentalization of cardiac beta(2)-adrenergic signaling

    J Biol Chem

    (1999)
  • A.L. Bauman et al.

    Dynamic regulation of cAMP synthesis through anchored PKA-adenylyl cyclase V/VI complexes

    Mol Cell

    (2006)
  • R. Efendiev et al.

    AKAP79 interacts with multiple adenylyl cyclase (AC) isoforms and scaffolds AC5 and -6 to alpha-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) receptors

    J Biol Chem

    (2010)
  • M.S. Kapiloff et al.

    An adenylyl cyclase-mAKAPbeta signaling complex regulates cAMP levels in cardiac myocytes

    J Biol Chem

    (2009)
  • D. Mika et al.

    PDEs create local domains of cAMP signaling

    J Mol Cell Cardiol

    (2012)
  • R.V. Iancu et al.

    Compartmentation of cAMP signaling in cardiac myocytes: a computational study

    Biophys J

    (2007)
  • K.A. Tasken et al.

    Phosphodiesterase 4D and protein kinase a type II constitute a signaling unit in the centrosomal area

    J Biol Chem

    (2001)
  • M. Conti et al.

    Cyclic AMP-specific PDE4 phosphodiesterases as critical components of cyclic AMP signaling

    J Biol Chem

    (2003)
  • S.E. Lehnart et al.

    Phosphodiesterase 4D deficiency in the ryanodine-receptor complex promotes heart failure and arrhythmias

    Cell

    (2005)
  • A. Yoshida et al.

    Cyclic AMP-dependent phosphorylation and regulation of the cardiac dihydropyridine-sensitive Ca channel

    FEBS Lett

    (1992)
  • Y. Fu et al.

    Deletion of the distal C terminus of CaV1.2 channels leads to loss of beta-adrenergic regulation and heart failure in vivo

    J Biol Chem

    (2011)
  • X. Wei et al.

    Modification of Ca2 + channel activity by deletions at the carboxyl terminus of the cardiac α1 subunit

    J Biol Chem

    (1994)
  • T. Lemke et al.

    Unchanged beta-adrenergic stimulation of cardiac L-type calcium channels in Ca v 1.2 phosphorylation site S1928A mutant mice

    J Biol Chem

    (2008)
  • T. Gao et al.

    cAMP-dependent regulation of cardiac L-type Ca2 + channels requires membrane targeting of PKA and phosphorylation of channel subunits

    Neuron

    (1997)
  • S.F. Oliveria et al.

    AKAP79/150 anchoring of calcineurin controls neuronal L-type Ca2 + channel activity and nuclear signaling

    Neuron

    (2007)
  • M. Bunemann et al.

    Functional regulation of L-type calcium channels via protein kinase A-mediated phosphorylation of the beta(2) subunit

    J Biol Chem

    (1999)
  • H. Haase et al.

    Phosphorylation of the L-type calcium channel beta subunit is involved in beta-adrenergic signal transduction in canine myocardium

    FEBS Lett

    (1993)
  • S. Dai et al.

    Supramolecular assemblies and localized regulation of voltage-gated ion channels

    Physiol Rev

    (2009)
  • E. Carafoli

    Calcium signaling: a tale for all seasons

    Proc Natl Acad Sci U S A

    (2002)
  • D.M. Bers

    Cardiac excitation–contraction coupling

    Nature

    (2002)
  • W.A. Catterall

    Structure and regulation of voltage-gated Ca2 + channels

    Annu Rev Cell Dev Biol

    (2000)
  • D.M. Bers et al.

    Ca channels in cardiac myocytes: structure and function in Ca influx and intracellular Ca release

    Cardiovasc Res

    (1999)
  • J.T. Hulme et al.

    Autoinhibitory control of the CaV1.2 channel by its proteolytically processed distal C-terminal domain

    J Physiol

    (2006)
  • G. Mikala et al.

    cAMP-dependent phosphorylation sites and macroscopic activity of recombinant cardiac L-type calcium channels

    Mol Cell Biochem

    (1998)
  • K.S. De Jongh et al.

    Specific phosphorylation of a site in the full-length form of the alpha 1 subunit of the cardiac L-type calcium channel by adenosine 3′,5′-cyclic monophosphate-dependent protein kinase

    Biochemistry

    (1996)
  • J.D. Foell et al.

    Molecular heterogeneity of calcium channel beta-subunits in canine and human heart: evidence for differential subcellular localization

    Physiol Genomics

    (2004)
  • D. Singer et al.

    The roles of the subunits in the function of the calcium channel

    Science

    (1991)
  • T.J. Kamp et al.

    Enhancement of ionic current and charge movement by coexpression of calcium channel beta 1A subunit with alpha 1C subunit in a human embryonic kidney cell line

    J Physiol

    (1996)
  • R. Bangalore et al.

    Influence of L-type Ca channel alpha 2/delta-subunit on ionic and gating current in transiently transfected HEK 293 cells

    Am J Physiol

    (1996)
  • A.C. Dolphin

    Calcium channel auxiliary alpha(2)delta and beta subunits: trafficking and one step beyond

    Nat Rev Neurosci

    (2012)
  • Cited by (66)

    View all citing articles on Scopus
    View full text